Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Drug Metab Dispos ; 46(3): 237-247, 2018 03.
Article in English | MEDLINE | ID: mdl-29311136

ABSTRACT

Momelotinib (MMB), a small-molecule inhibitor of Janus kinase (JAK)1/2 and of activin A receptor type 1 (ACVR1), is in clinical development for the treatment of myeloproliferative neoplasms. The pharmacokinetics and disposition of [14C]MMB were characterized in a single-dose, human mass-balance study. Metabolism and the pharmacologic activity of key metabolites were elucidated in multiple in vitro and in vivo experiments. MMB was rapidly absorbed following oral dosing with approximately 97% of the radioactivity recovered, primarily in feces with urine as a secondary route. Mean blood-to-plasma [14C] area under the plasma concentration-time curve ratio was 0.72, suggesting low association of MMB and metabolites with blood cells. [14C]MMB-derived radioactivity was detectable in blood for ≤48 hours, suggesting no irreversible binding of MMB or its metabolites. The major circulating human metabolite, M21 (a morpholino lactam), is a potent inhibitor of JAK1/2 and ACVR1 in vitro. Estimation of pharmacological activity index suggests M21 contributes significantly to the pharmacological activity of MMB for the inhibition of both JAK1/2 and ACVR1. M21 was observed in disproportionately higher amounts in human plasma than in rat or dog, the rodent and nonrodent species used for the general nonclinical safety assessment of this molecule. This discrepancy was resolved with additional nonclinical studies wherein the circulating metabolites and drug-drug interactions were further characterized. The human metabolism of MMB was mediated primarily by multiple cytochrome P450 enzymes, whereas M21 formation involved initial P450 oxidation of the morpholine ring followed by metabolism via aldehyde oxidase.


Subject(s)
Benzamides/pharmacokinetics , Pyrimidines/pharmacokinetics , Adolescent , Adult , Animals , Cell Line , Cell Line, Tumor , Cytochrome P-450 Enzyme System/metabolism , Dogs , Drug Interactions/physiology , Female , Hep G2 Cells , Humans , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Rats , Young Adult
2.
Blood ; 129(13): 1823-1830, 2017 03 30.
Article in English | MEDLINE | ID: mdl-28188131

ABSTRACT

Patients with myelofibrosis (MF) often develop anemia and frequently become dependent on red blood cell transfusions. Results from a phase 2 study for the treatment of MF with the Janus kinase 1/2 (JAK1/2) inhibitor momelotinib (MMB) demonstrated that MMB treatment ameliorated anemia, which was unexpected for a JAK1/2 inhibitor, because erythropoietin-mediated JAK2 signaling is essential for erythropoiesis. Using a rat model of anemia of chronic disease, we demonstrated that MMB treatment can normalize hemoglobin and red blood cell numbers. We found that this positive effect is driven by direct inhibition of the bone morphogenic protein receptor kinase activin A receptor, type I (ACVR1), and the subsequent reduction of hepatocyte hepcidin production. Of note, ruxolitinib, a JAK1/2 inhibitor approved for the treatment of MF, had no inhibitory activity on this pathway. Further, we demonstrated the effect of MMB is not mediated by direct inhibition of JAK2-mediated ferroportin (FPN1) degradation, because neither MMB treatment nor myeloid-specific deletion of JAK2 affected FPN1 expression. Our data support the hypothesis that the improvement of inflammatory anemia by MMB results from inhibition of ACVR1-mediated hepcidin expression in the liver, which leads to increased mobilization of sequestered iron from cellular stores and subsequent stimulation of erythropoiesis.


Subject(s)
Anemia/drug therapy , Benzamides/therapeutic use , Bone Morphogenetic Protein Receptors, Type I/antagonists & inhibitors , Hepcidins/biosynthesis , Pyrimidines/therapeutic use , Activin Receptors, Type I/antagonists & inhibitors , Animals , Benzamides/pharmacology , Chronic Disease , Hepatocytes/metabolism , Iron/metabolism , Primary Myelofibrosis/complications , Pyrimidines/pharmacology , Rats
3.
Drug Metab Dispos ; 45(1): 76-85, 2017 01.
Article in English | MEDLINE | ID: mdl-27803021

ABSTRACT

Inhibition of thiamine transporters has been proposed as a putative mechanism for the observation of Wernicke's encephalopathy and subsequent termination of clinical development of fedratinib, a Janus kinase inhibitor (JAKi). This study aimed to determine the potential for other JAKi to inhibit thiamine transport using human epithelial colorectal adenocarcinoma (Caco-2) and thiamine transporter (THTR) overexpressing cells and to better elucidate the structural basis for interacting with THTR. Only JAKi containing a 2,4-diaminopyrimidine were observed to inhibit thiamine transporters. Fedratinib inhibited thiamine uptake into Caco-2 cells (IC50 = 0.940 µM) and THTR-2 (IC50 = 1.36 µM) and, to a lesser extent, THTR-1 (IC50 = 7.10 µM) overexpressing cells. Two other JAKi containing this moiety, AZD1480 and cerdulatinib, were weaker inhibitors of the thiamine transporters. Other JAKi-including monoaminopyrimidines, such as momelotinib, and nonaminopyrimidines, such as filgotinib-did not have any inhibitory effects on thiamine transport. A pharmacophore model derived from the minimized structure of thiamine suggests that 2,4-diaminopyrimidine-containing compounds can adopt a conformation matching several key features of thiamine. Further studies with drugs containing a 2,4-diaminopyrimidine resulted in the discovery that the antibiotic trimethoprim also potently inhibits thiamine uptake mediated by THTR-1 (IC50 = 6.84 µM) and THTR-2 (IC50 = 5.56 µM). Fedratinib and trimethoprim were also found to be substrates for THTR, a finding with important implications for their disposition in the body. In summary, our results show that not all JAKi have the potential to inhibit thiamine transport and further establish the interaction of these transporters with xenobiotics.


Subject(s)
Janus Kinase Inhibitors/pharmacology , Membrane Transport Proteins/metabolism , Pyrimidines/chemistry , Pyrrolidines/pharmacology , Sulfonamides/pharmacology , Trimethoprim/pharmacology , Caco-2 Cells , Drug Interactions , HEK293 Cells , Humans , Janus Kinase Inhibitors/chemistry , Membrane Transport Proteins/genetics , Molecular Structure , Pyrrolidines/chemistry , Substrate Specificity , Sulfonamides/chemistry , Thiamine/metabolism , Trimethoprim/chemistry
4.
J Med Chem ; 57(9): 3856-73, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24779514

ABSTRACT

Spleen tyrosine kinase (Syk) is an attractive drug target in autoimmune, inflammatory, and oncology disease indications. The most advanced Syk inhibitor, R406, 1 (or its prodrug form fostamatinib, 2), has shown efficacy in multiple therapeutic indications, but its clinical progress has been hampered by dose-limiting adverse effects that have been attributed, at least in part, to the off-target activities of 1. It is expected that a more selective Syk inhibitor would provide a greater therapeutic window. Herein we report the discovery and optimization of a novel series of imidazo[1,2-a]pyrazine Syk inhibitors. This work culminated in the identification of GS-9973, 68, a highly selective and orally efficacious Syk inhibitor which is currently undergoing clinical evaluation for autoimmune and oncology indications.


Subject(s)
Indazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazines/pharmacology , Spleen/drug effects , Administration, Oral , Animals , Cells, Cultured , Drug Discovery , Humans , Indazoles/administration & dosage , Indazoles/chemistry , Magnetic Resonance Spectroscopy , Mice , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Pyrazines/administration & dosage , Pyrazines/chemistry , Rats , Spectrometry, Mass, Electrospray Ionization , Spleen/enzymology , Structure-Activity Relationship
5.
Nat Chem Biol ; 7(1): 41-50, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21113169

ABSTRACT

Bruton's tyrosine kinase (Btk) is a therapeutic target for rheumatoid arthritis, but the cellular and molecular mechanisms by which Btk mediates inflammation are poorly understood. Here we describe the discovery of CGI1746, a small-molecule Btk inhibitor chemotype with a new binding mode that stabilizes an inactive nonphosphorylated enzyme conformation. CGI1746 has exquisite selectivity for Btk and inhibits both auto- and transphosphorylation steps necessary for enzyme activation. Using CGI1746, we demonstrate that Btk regulates inflammatory arthritis by two distinct mechanisms. CGI1746 blocks B cell receptor-dependent B cell proliferation and in prophylactic regimens reduces autoantibody levels in collagen-induced arthritis. In macrophages, Btk inhibition abolishes FcγRIII-induced TNFα, IL-1ß and IL-6 production. Accordingly, in myeloid- and FcγR-dependent autoantibody-induced arthritis, CGI1746 decreases cytokine levels within joints and ameliorates disease. These results provide new understanding of the function of Btk in both B cell- or myeloid cell-driven disease processes and provide a compelling rationale for targeting Btk in rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , B-Lymphocytes/drug effects , Benzamides/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Myeloid Cells/drug effects , Protein Kinase Inhibitors/therapeutic use , Agammaglobulinaemia Tyrosine Kinase , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Autoantibodies/immunology , Autoantibodies/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Benzamides/chemistry , Benzamides/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Mice , Myeloid Cells/immunology , Myeloid Cells/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/pharmacology , Protein-Tyrosine Kinases/therapeutic use , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
6.
Immunity ; 27(4): 610-24, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17936032

ABSTRACT

The maturation of dendritic cells (DCs) after exposure to microbial products or inflammatory mediators plays a critical role in initiating the immune response. We found that maturation can also occur under steady-state conditions, triggered by alterations in E-cadherin-mediated DC-DC adhesion. Selective disruption of these interactions induced the typical features of DC maturation including the upregulation of costimulatory molecules, MHC class II, and chemokine receptors. These events were triggered at least in part by activation of the beta-catenin pathway. However, unlike maturation induced by microbial products, E-cadherin-stimulated DCs failed to release immunostimulatory cytokines, exhibiting an entirely different transcriptional profile. As a result, E-cadherin-stimulated DCs elicited an entirely different T cell response in vivo, generating T cells with a regulatory as opposed to an effector phenotype. These DCs induced tolerance in vivo and may thus contribute to the elusive steady-state "tolerogenic DCs."


Subject(s)
Cadherins/metabolism , Cell Adhesion/immunology , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Adoptive Transfer , Animals , Antigen Presentation/immunology , Blotting, Western , Cadherins/immunology , Cell Communication/immunology , Cells, Cultured , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression , Gene Expression Regulation , Humans , Immune Tolerance/physiology , Immunoprecipitation , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Protein Array Analysis , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , TCF Transcription Factors/immunology , TCF Transcription Factors/metabolism , beta Catenin/immunology , beta Catenin/metabolism
7.
Assay Drug Dev Technol ; 5(1): 49-64, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17355199

ABSTRACT

The importance of protein kinases as a major class of drug targets across multiple diseases has generated a critical need for technologies that enable the identification of potent and selective kinase inhibitors. Bruton's tyrosine kinase (Btk) is a compelling drug target in multiple therapeutic areas, including systemic lupus erythematosus, asthma, rheumatoid arthritis, and B cell malignancies. We have combined potent, selective kinase inhibition through chemical genetics with gene expression profiling to identify a "fingerprint" of transcriptional changes associated with selective Btk kinase inhibition. The Btk transcriptional fingerprint shows remarkable relevance for Btk's biological roles and was used for functional selectivity profiling of two kinase inhibitor compounds. The fingerprint was able to rank the compounds by relative selectivity for Btk, and revealed broader off-target effects than observed in a broad panel of biochemical kinase cross screens. In addition to being useful for functional selectivity profiling, the fingerprint genes are themselves potential preclinical and clinical biomarkers for developing Btk-directed therapies.


Subject(s)
Gene Expression Profiling/methods , Kidney/metabolism , Peptide Mapping/methods , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/analysis , Protein Kinases/metabolism , Transcription Factors/metabolism , Biological Assay/methods , Cell Line , Humans , Kidney/drug effects , Oligonucleotide Array Sequence Analysis/methods , Protein Kinases/genetics , Transcription Factors/genetics
8.
Assay Drug Dev Technol ; 2(4): 417-29, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15357923

ABSTRACT

Protein kinases play key roles in a number of diseases, including cancer, inflammation, and diabetes. Disregulation of kinase-based signal transduction networks results in aberrant cell differentiation, activation, proliferation, and invasion. The growing importance of kinases as a major class of drug targets across multiple large clinical indications, together with the large number of kinases in the genome (~518), has generated a critical need for technologies that enable the identification of potent and selective kinase inhibitors with good drug-like properties. In this review, we describe methods used for developing cell-based assays for kinase inhibitors, discuss advantages and disadvantages of each approach, and describe new chemical genetic methods as reference systems for establishing cell-based assays and their use for functional selectivity profiling of kinase inhibitors.


Subject(s)
Protein Kinase Inhibitors/analysis , Protein Kinases/genetics , Protein Kinases/metabolism , Technology, Pharmaceutical/methods , Amino Acid Sequence , Molecular Sequence Data , Protein Kinase Inhibitors/standards , Reference Standards , Reference Values
SELECTION OF CITATIONS
SEARCH DETAIL
...